Register      Login
Reproduction, Fertility and Development Reproduction, Fertility and Development Society
Vertebrate reproductive science and technology
REVIEW

Current knowledge and the future potential of extracellular vesicles in mammalian reproduction

Dawit Tesfaye https://orcid.org/0000-0001-8166-0606 A * , Nico Menjivar A and Samuel Gebremedhn B
+ Author Affiliations
- Author Affiliations

A Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, 3051 Rampart Road, Fort Collins, CO 80521, USA.

B Genus plc, 1525 River Road, DeForest, WI 53532, USA.

* Correspondence to: Dawit.Tesfaye@Colostate.edu

Reproduction, Fertility and Development 34(2) 174-189 https://doi.org/10.1071/RD21277
Published online: 12 October 2021

© 2022 The Author(s) (or their employer(s)). Published by CSIRO Publishing on behalf of the IETS

Abstract

Extracellular vesicles (EVs), which contain various functional classes of vesicles, namely exosomes, microvesicles, and apoptotic bodies, represent the major nano-shuttle to transfer bioactive molecules from donor to recipient cells to facilitate cell-to-cell communication in the follicular, oviduct, and uterine microenvironments. In addition to transferring various molecular cargos in the form of miRNAs, mRNAs, proteins, lipids, and DNA molecules, the relative proportion of those molecular cargos in the reproductive fluids can be associated with the physiological and pathological condition of the host animal. Inside the follicle, EV-mediated circulation of miRNAs has been reported to be associated with the growth status of the enclosed oocytes, the metabolic status, and the advanced maternal aging of the animal. Importantly, EVs have the potential to protect their cargo molecules from extracellular degradation or modification while travelling to the recipient cells. This fact together with the enormous availability in almost all biological fluids and spent culture media make them attractive in the search for biomarkers of oocyte/embryo developmental competence, receptive maternal environment and a multitude of reproductive pathophysiological conditions. One of the key factors that have contributed to the lower efficiency of assisted reproductive technologies (ART) is the absence of several maternal in vivo factors in the ART procedures. For this, several studies have been conducted to supplement various components present in the follicular and oviductal fluids into the existing ART procedures and significant positive impacts have been observed in terms of embryo cleavage rate, blastocyst rate, resistance to stress, and survival after cryopreservation. The potential of EVs in shuttling protective messages against environmental and physiological stressors has been evidenced. The effective use of the EV-coupled molecular signals against stress-associated conditions has the potential to pave the path for the application of these protective signals against oxidative stress-associated pathological conditions including PCOS, ageing, and endometritis. In this review, we provide current knowledge and potential future use of EVs as remedies in reproductive pathophysiological conditions, mainly in follicular and oviductal microenvironments.

Keywords: assisted reproductive technology, DNA, extracellular vesicles, reproduction, RNA, stress response.


References

Agarwal, A, Aponte-Mellado, A, Premkumar, BJ, Shaman, A, and Gupta, S (2012). The effects of oxidative stress on female reproduction: a review. Reproductive Biology and Endocrinology 10, 49.
The effects of oxidative stress on female reproduction: a review.Crossref | GoogleScholarGoogle Scholar | 22748101PubMed |

Al-Dossary, AA, Strehler Emanuel, E, and Martin-Deleon, PA (2013). Expression and secretion of plasma membrane Ca2+-ATPase 4a (PMCA4a) during murine estrus: association with oviductal exosomes and uptake in sperm. PLoS ONE 8, e80181.
Expression and secretion of plasma membrane Ca2+-ATPase 4a (PMCA4a) during murine estrus: association with oviductal exosomes and uptake in sperm.Crossref | GoogleScholarGoogle Scholar | 24244642PubMed |

Alemu, TW, Pandey, HO, Salilew Wondim, D, Gebremedhn, S, Neuhof, C, Tholen, E, et al. (2018). Oxidative and endoplasmic reticulum stress defense mechanisms of bovine granulosa cells exposed to heat stress. Theriogenology 110, 130–141.
Oxidative and endoplasmic reticulum stress defense mechanisms of bovine granulosa cells exposed to heat stress.Crossref | GoogleScholarGoogle Scholar | 29396041PubMed |

Almagor, M, Bejar, C, Kafka, I, and Yaffe, H (1996). Pregnancy rates after communal growth of preimplantation human embryos in vitro. Fertility and Sterility 66, 394–397.
Pregnancy rates after communal growth of preimplantation human embryos in vitro.Crossref | GoogleScholarGoogle Scholar | 8751736PubMed |

Almiñana, C, Corbin, E, Tsikis, G, Alcântara-Neto, AS, Labas, V, Reynaud, K, et al. (2017). Oviduct extracellular vesicles protein content and their role during oviduct-embryo cross-talk. Reproduction (Cambridge, England) 154, 153–168.
Oviduct extracellular vesicles protein content and their role during oviduct-embryo cross-talk.Crossref | GoogleScholarGoogle Scholar |

Almiñana, C, Tsikis, G, Labas, V, Uzbekov, R, da Silveira, JC, Bauersachs, S, and Mermillod, P (2018). Deciphering the oviductal extracellular vesicles content across the estrous cycle: implications for the gametes–oviduct interactions and the environment of the potential embryo. BMC Genomics 19, 622.
Deciphering the oviductal extracellular vesicles content across the estrous cycle: implications for the gametes–oviduct interactions and the environment of the potential embryo.Crossref | GoogleScholarGoogle Scholar | 30134841PubMed |

Alvarez-Erviti, L, Seow, Y, Yin, H, Betts, C, Lakhal, S, and Wood, MJA (2011). Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nature Biotechnology 29, 341–345.
Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes.Crossref | GoogleScholarGoogle Scholar | 21423189PubMed |

Aprioku, JS (2013). Pharmacology of free radicals and the impact of reactive oxygen species on the testis. Journal of Reproduction & Infertility 14, 158–172.

Asaadi, A, Dolatabad, NA, Atashi, H, Raes, A, van Damme, P, Hoelker, M, et al. (2021). Extracellular vesicles from follicular and ampullary fluid isolated by density gradient ultracentrifugation improve bovine embryo development and quality. International Journal of Molecular Sciences 22, 578.
Extracellular vesicles from follicular and ampullary fluid isolated by density gradient ultracentrifugation improve bovine embryo development and quality.Crossref | GoogleScholarGoogle Scholar |

Avilés, M, Gutiérrez-Adán, A, and Coy, P (2010). Oviductal secretions: will they be key factors for the future ARTs? In Molecular Human Reproduction 16, 896–906.
Oviductal secretions: will they be key factors for the future ARTs?Crossref | GoogleScholarGoogle Scholar |

Baddela, VS, Sharma, A, Viergutz, T, Koczan, D, and Vanselow, J (2018). Low oxygen levels induce early luteinization associated changes in bovine granulosa cells. Frontiers in Physiology 9, 1066.
Low oxygen levels induce early luteinization associated changes in bovine granulosa cells.Crossref | GoogleScholarGoogle Scholar | 30131718PubMed |

Baixauli, F, López-Otín, C, and Mittelbrunn, M (2014). Exosomes and autophagy: coordinated mechanisms for the maintenance of cellular fitness. Frontiers in Immunology 5, 403.
Exosomes and autophagy: coordinated mechanisms for the maintenance of cellular fitness.Crossref | GoogleScholarGoogle Scholar | 25191326PubMed |

Beam, SW, and Butler, WR (1997). Energy balance and ovarian follicle development prior to the first ovulation postpartum in dairy cows receiving three levels of dietary fat. Biology of Reproduction 56, 133–142.
Energy balance and ovarian follicle development prior to the first ovulation postpartum in dairy cows receiving three levels of dietary fat.Crossref | GoogleScholarGoogle Scholar | 9002642PubMed |

Beam, SW, and Butler, WR (1999). Effects of energy balance on follicular development and first ovulation in postpartum dairy cows. Journal of Reproduction and Fertility 54, 411–424.
| 10692872PubMed |

Belleannée, C, Calvo, É, Caballero, J, and Sullivan, R (2013). Epididymosomes convey different repertoires of microRNAs throughout the bovine epididymis. Biology of Reproduction 89, 30.
Epididymosomes convey different repertoires of microRNAs throughout the bovine epididymis.Crossref | GoogleScholarGoogle Scholar | 23803555PubMed |

Bewicke-Copley, F, Mulcahy, LA, Jacobs, LA, Samuel, P, Akbar, N, Pink, RC, and Carter, DRF (2017). Extracellular vesicles released following heat stress induce bystander effect in unstressed populations. Journal of Extracellular Vesicles 6, 1340746.
Extracellular vesicles released following heat stress induce bystander effect in unstressed populations.Crossref | GoogleScholarGoogle Scholar | 28717426PubMed |

Bosco, D, Haefliger, J-A, and Meda, P (2011). Connexins: key mediators of endocrine function. Physiological Reviews 91, 1393–1445.
Connexins: key mediators of endocrine function.Crossref | GoogleScholarGoogle Scholar | 22013215PubMed |

Brower, PT, and Schultz, RM (1982). Intercellular communication between granulosa cells and mouse oocytes: existence and possible nutritional role during oocyte growth. Developmental Biology 90, 144–153.
Intercellular communication between granulosa cells and mouse oocytes: existence and possible nutritional role during oocyte growth.Crossref | GoogleScholarGoogle Scholar | 7199496PubMed |

Burton, GJ, and Jauniaux, E (2011). Oxidative stress. Best Practice & Research Clinical Obstetrics & Gynaecology 25, 287–299.
Oxidative stress.Crossref | GoogleScholarGoogle Scholar |

Burton, KA, and McKnight, GS (2007). PKA, germ cells, and fertility. Physiology (Bethesda, MD) 22, 40–46.
PKA, germ cells, and fertility.Crossref | GoogleScholarGoogle Scholar |

Burns, G, Brooks, K, Wildung, M, Navakanitworakul, R, Christenson, LK, and Spencer, TE (2014). Extracellular vesicles in luminal fluid of the ovine uterus. PLoS ONE 9, e90913.
Extracellular vesicles in luminal fluid of the ovine uterus.Crossref | GoogleScholarGoogle Scholar | 24614226PubMed |

Burns, GW, Brooks, KE, and Spencer, TE (2016). Extracellular vesicles originate from the conceptus and uterus during early pregnancy in sheep. Biology of Reproduction 94, 56.
Extracellular vesicles originate from the conceptus and uterus during early pregnancy in sheep.Crossref | GoogleScholarGoogle Scholar | 26819476PubMed |

Burns, GW, Brooks, KE, O’Neil, EV, Hagen, DE, Behura, SK, and Spencer, TE (2018). Progesterone effects on extracellular vesicles in the sheep uterus. Biology of Reproduction 98, 612–622.
Progesterone effects on extracellular vesicles in the sheep uterus.Crossref | GoogleScholarGoogle Scholar | 29346527PubMed |

Can, U, Buyukinan, M, and Yerlikaya, FH (2016). The investigation of circulating microRNAs associated with lipid metabolism in childhood obesity. Pediatric Obesity 11, 228–234.
The investigation of circulating microRNAs associated with lipid metabolism in childhood obesity.Crossref | GoogleScholarGoogle Scholar | 26223376PubMed |

Castaño, C, Kalko, S, Novials, A, and Párrizas, M (2018). Obesity-associated exosomal miRNAs modulate glucose and lipid metabolism in mice. Proceedings of the National Academy of Sciences of the United States of America 115, 12158–12163.
Obesity-associated exosomal miRNAs modulate glucose and lipid metabolism in mice.Crossref | GoogleScholarGoogle Scholar | 30429322PubMed |

Chairoungdua, A, Smith, DL, Pochard, P, Hull, M, and Caplan, MJ (2010). Exosome release of β-catenin: a novel mechanism that antagonizes Wnt signaling. The Journal of Cell Biology 190, 1079–1091.
Exosome release of β-catenin: a novel mechanism that antagonizes Wnt signaling.Crossref | GoogleScholarGoogle Scholar | 20837771PubMed |

Che, X, Jian, F, Chen, C, Liu, C, Liu, G, and Feng, W (2020). PCOS serum-derived exosomal miR-27a-5p stimulates endometrial cancer cells migration and invasion. Journal of Molecular Endocrinology 64, 1–12.
PCOS serum-derived exosomal miR-27a-5p stimulates endometrial cancer cells migration and invasion.Crossref | GoogleScholarGoogle Scholar | 31710594PubMed |

Cheng, L, Doecke, JD, Sharples, RA, Villemagne, VL, Fowler, CJ, Rembach, A, et al. (2015). Prognostic serum miRNA biomarkers associated with Alzheimer’s disease shows concordance with neuropsychological and neuroimaging assessment. Molecular Psychiatry 20, 1188–1196.
Prognostic serum miRNA biomarkers associated with Alzheimer’s disease shows concordance with neuropsychological and neuroimaging assessment.Crossref | GoogleScholarGoogle Scholar | 25349172PubMed |

Colombo, M, Raposo, G, and Théry, C (2014). Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annual Review of Cell and Developmental Biology 30, 255–289.
Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles.Crossref | GoogleScholarGoogle Scholar | 25288114PubMed |

Conti, M, Hsieh, M, Zamah, AM, and Oh, JS (2012). Novel signaling mechanisms in the ovary during oocyte maturation and ovulation. Molecular and Cellular Endocrinology 356, 65–73.
Novel signaling mechanisms in the ovary during oocyte maturation and ovulation.Crossref | GoogleScholarGoogle Scholar | 22101318PubMed |

Contreras, GA, and Sordillo, LM (2011). Lipid mobilization and inflammatory responses during the transition period of dairy cows. Comparative Immunology, Microbiology and Infectious Diseases 34, 281–289.
Lipid mobilization and inflammatory responses during the transition period of dairy cows.Crossref | GoogleScholarGoogle Scholar | 21316109PubMed |

Coy, P, García-Vázquez, FA, Visconti, PE, and Avilés, M (2012). Roles of the oviduct in mammalian fertilization. Reproduction (Cambridge, England) 144, 649–660.
Roles of the oviduct in mammalian fertilization.Crossref | GoogleScholarGoogle Scholar |

Cronqvist, T, Erlandsson, L, Tannetta, D, and Hansson, SR (2020). Placental syncytiotrophoblast extracellular vesicles enter primary endothelial cells through clathrin-mediated endocytosis. Placenta 100, 133–141.
Placental syncytiotrophoblast extracellular vesicles enter primary endothelial cells through clathrin-mediated endocytosis.Crossref | GoogleScholarGoogle Scholar | 32980046PubMed |

Cruz, L, Romero, JAA, Iglesia, RP, and Lopes, MH (2018). Extracellular vesicles: decoding a new language for cellular communication in early embryonic Development. Frontiers in Cell and Developmental Biology 6, 94.
Extracellular vesicles: decoding a new language for cellular communication in early embryonic Development.Crossref | GoogleScholarGoogle Scholar | 30211159PubMed |

Da Silveira, JC, Veeramachaneni, DNR, Winger, QA, Carnevale, EM, and Bouma, GJ (2012). Cell-secreted vesicles in equine ovarian follicular fluid contain miRNAs and proteins: a possible new form of cell communication within the ovarian follicle. Biology of Reproduction 86, 71.
Cell-secreted vesicles in equine ovarian follicular fluid contain miRNAs and proteins: a possible new form of cell communication within the ovarian follicle.Crossref | GoogleScholarGoogle Scholar | 22116803PubMed |

de Maio, A (2011). Extracellular heat shock proteins, cellular export vesicles, and the Stress Observation System: a form of communication during injury, infection, and cell damage. It is never known how far a controversial finding will go! Dedicated to Ferruccio Ritossa. Cell Stress & Chaperones 16, 235–249.
Extracellular heat shock proteins, cellular export vesicles, and the Stress Observation System: a form of communication during injury, infection, and cell damage. It is never known how far a controversial finding will go! Dedicated to Ferruccio Ritossa.Crossref | GoogleScholarGoogle Scholar |

Dhahbi, JM, Spindler, SR, Atamna, H, Yamakawa, A, Guerrero, N, Boffelli, D, et al. (2013). Deep sequencing identifies circulating mouse miRNAs that are functionally implicated in manifestations of aging and responsive to calorie restriction. Aging 5, 130–141.
Deep sequencing identifies circulating mouse miRNAs that are functionally implicated in manifestations of aging and responsive to calorie restriction.Crossref | GoogleScholarGoogle Scholar | 23470454PubMed |

Diez-Fraile, A, Lammens, T, Tilleman, K, Witkowski, W, Verhasselt, B, de Sutter, Petra, et al. (2014). Age-associated differential microRNA levels in human follicular fluid reveal pathways potentially determining fertility and success of in vitro fertilization. Human Fertility (Cambridge, England) 17, 90–98.
Age-associated differential microRNA levels in human follicular fluid reveal pathways potentially determining fertility and success of in vitro fertilization.Crossref | GoogleScholarGoogle Scholar |

Dong, H, Wang, C, Lu, S, Yu, C, Huang, L, Feng, W, et al. (2016). A panel of four decreased serum microRNAs as a novel biomarker for early Parkinson’s disease. Biomarkers: Biochemical Indicators of Exposure, Response, and Susceptibility to Chemicals 21, 129–137.
A panel of four decreased serum microRNAs as a novel biomarker for early Parkinson’s disease.Crossref | GoogleScholarGoogle Scholar |

Donnay, I, van Langendonckt, A, Auquier, P, Grisart, B, Vansteenbrugge, A, Massip, A, and Dessy, F (1997). Effects of co-culture and embryo number on the in vitro development of bovine embryos. Theriogenology 47, 1549–1561.
Effects of co-culture and embryo number on the in vitro development of bovine embryos.Crossref | GoogleScholarGoogle Scholar | 16728097PubMed |

Ebner, T, Shebl, O, Moser, M, Mayer, RB, Arzt, W, and Tews, G (2010). Group culture of human zygotes is superior to individual culture in terms of blastulation, implantation and life birth. Reproductive Biomedicine Online 21, 762–768.
Group culture of human zygotes is superior to individual culture in terms of blastulation, implantation and life birth.Crossref | GoogleScholarGoogle Scholar | 21051291PubMed |

Edwards, JL, and Hansen, PJ (1996). Elevated temperature increases heat shock protein 70 synthesis in bovine two-cell embryos and compromises function of maturing oocytes. Biology of Reproduction 55, 341–346.
Elevated temperature increases heat shock protein 70 synthesis in bovine two-cell embryos and compromises function of maturing oocytes.Crossref | GoogleScholarGoogle Scholar | 8828838PubMed |

Eldh, M, Ekström, K, Valadi, H, Sjöstrand, M, Olsson, B, Jernås, M, and Lötvall, J (2010). Exosomes communicate protective messages during oxidative stress; possible role of exosomal shuttle RNA. PLoS ONE 5, e15353.
Exosomes communicate protective messages during oxidative stress; possible role of exosomal shuttle RNA.Crossref | GoogleScholarGoogle Scholar | 21179422PubMed |

Escrevente, C, Keller, S, Altevogt, P, and Costa, J (2011). Interaction and uptake of exosomes by ovarian cancer cells. BMC Cancer 11, 108.
Interaction and uptake of exosomes by ovarian cancer cells.Crossref | GoogleScholarGoogle Scholar | 21439085PubMed |

Fabbri, M, Paone, A, Calore, F, Galli, R, Gaudio, E, Santhanam, R, et al. (2012). MicroRNAs bind to Toll-like receptors to induce prometastatic inflammatory response. Proceedings of the National Academy of Sciences of the United States of America 109, E2110–E2116.
MicroRNAs bind to Toll-like receptors to induce prometastatic inflammatory response.Crossref | GoogleScholarGoogle Scholar | 22753494PubMed |

Fazeli, A (2011). Maternal communication with gametes and embryo: a personal opinion. Reproduction in Domestic Animals = Zuchthygiene 46, 75–78.
Maternal communication with gametes and embryo: a personal opinion.Crossref | GoogleScholarGoogle Scholar | 21884283PubMed |

Fereshteh, Z, Bathala, P, Galileo, DS, and Martin-Deleon, PA (2019). Detection of extracellular vesicles in the mouse vaginal fluid: their delivery of sperm proteins that stimulate capacitation and modulate fertility. Journal of Cellular Physiology 234, 12745–12756.
Detection of extracellular vesicles in the mouse vaginal fluid: their delivery of sperm proteins that stimulate capacitation and modulate fertility.Crossref | GoogleScholarGoogle Scholar | 30536802PubMed |

Fortune, JE (1994). Ovarian follicular growth and development in mammals. Biology of Reproduction 50, 225–232.
Ovarian follicular growth and development in mammals.Crossref | GoogleScholarGoogle Scholar | 8142540PubMed |

Fragouli, E, Alfarawati, S, Spath, K, and Wells, D (2014). Morphological and cytogenetic assessment of cleavage and blastocyst stage embryos. Molecular Human Reproduction 20, 117–126.
Morphological and cytogenetic assessment of cleavage and blastocyst stage embryos.Crossref | GoogleScholarGoogle Scholar | 24184690PubMed |

Franchi, A, Cubilla, M, Guidobaldi, HA, Bravo, AA, and Giojalas, LC (2016). Uterosome-like vesicles prompt human sperm fertilizing capability. Molecular Human Reproduction 22, 833–841.
Uterosome-like vesicles prompt human sperm fertilizing capability.Crossref | GoogleScholarGoogle Scholar | 27678485PubMed |

Galimberti, D, Villa, C, Fenoglio, C, Serpente, M, Ghezzi, L, Cioffi, SMG, et al. (2014). Circulating miRNAs as potential biomarkers in Alzheimer’s disease. Journal of Alzheimer’s Disease 42, 1261–1267.
Circulating miRNAs as potential biomarkers in Alzheimer’s disease.Crossref | GoogleScholarGoogle Scholar | 25024331PubMed |

Gatien, J, Mermillod, P, Tsikis, G, Bernardi, O, Janati Idrissi, S, Uzbekov, R, et al. (2019). Metabolomic profile of oviductal extracellular vesicles across the estrous cycle in cattle. International Journal of Molecular Sciences 20, .
Metabolomic profile of oviductal extracellular vesicles across the estrous cycle in cattle.Crossref | GoogleScholarGoogle Scholar | 31888194PubMed |

Gebremedhn, S, Ali, A, Gad, A, Prochazka, R, and Tesfaye, D (2020a). Extracellular vesicles as mediators of environmental and metabolic stress coping mechanisms during mammalian follicular development. Frontiers in Veterinary Science 7, 602043.
Extracellular vesicles as mediators of environmental and metabolic stress coping mechanisms during mammalian follicular development.Crossref | GoogleScholarGoogle Scholar | 33330723PubMed |

Gebremedhn, S, Gad, A, Aglan, HS, Laurincik, J, Prochazka, R, Salilew-Wondim, D, et al. (2020b). Extracellular vesicles shuttle protective messages against heat stress in bovine granulosa cells. Scientific Reports 10, 15824.
Extracellular vesicles shuttle protective messages against heat stress in bovine granulosa cells.Crossref | GoogleScholarGoogle Scholar | 32978452PubMed |

Golini, VE, Stradaioli, G, and Sirard, MA (2014). Transcriptome analysis of bovine granulosa cells of preovulatory follicles harvested 30, 60, 90, and 120 days postpartum. Theriogenology 82, 580–591.e5.
Transcriptome analysis of bovine granulosa cells of preovulatory follicles harvested 30, 60, 90, and 120 days postpartum.Crossref | GoogleScholarGoogle Scholar | 24985356PubMed |

Greening, DW, Nguyen, HPT, Elgass, K, Simpson Richard, J, and Salamonsen, LA (2016). Human endometrial exosomes contain hormone-specific cargo modulating trophoblast adhesive capacity: insights into endometrial–embryo interactions. Biology of Reproduction 94, 38.
Human endometrial exosomes contain hormone-specific cargo modulating trophoblast adhesive capacity: insights into endometrial–embryo interactions.Crossref | GoogleScholarGoogle Scholar | 26764347PubMed |

Hailay, T, Hoelker, M, Poirier, M, Gebremedhn, S, Rings, F, Saeed-Zidane, M, et al. (2019). Extracellular vesicle-coupled miRNA profiles in follicular fluid of cows with divergent post-calving metabolic status. Scientific Reports 9, 12851.
Extracellular vesicle-coupled miRNA profiles in follicular fluid of cows with divergent post-calving metabolic status.Crossref | GoogleScholarGoogle Scholar | 31492906PubMed |

Hamdi, M, Lopera-Vasquez, R, Maillo, V, Sanchez-Calabuig, MJ, Núnez, C, Gutierrez-Adan, A, and Rizos, D (2018). Bovine oviductal and uterine fluid support in vitro embryo development. Reproduction, Fertility, and Development 30, 935–945.
Bovine oviductal and uterine fluid support in vitro embryo development.Crossref | GoogleScholarGoogle Scholar | 29167013PubMed |

Hansen, LB (2000). Consequences of selection for milk yield from a geneticist’s viewpoint. Journal of Dairy Science 83, 1145–1150.
Consequences of selection for milk yield from a geneticist’s viewpoint.Crossref | GoogleScholarGoogle Scholar | 10821591PubMed |

Hanson, PI, and Cashikar, A (2012). Multivesicular body morphogenesis. Annual Review of Cell and Developmental Biology 28, 337–362.
Multivesicular body morphogenesis.Crossref | GoogleScholarGoogle Scholar | 22831642PubMed |

Harris, EA, Stephens, KK, and Winuthayanon, W (2020). Extracellular vesicles and the oviduct function. International Journal of Molecular Sciences 21, 8280.
Extracellular vesicles and the oviduct function.Crossref | GoogleScholarGoogle Scholar |

Heidari-Khoei, H, Esfandiari, F, Hajari, MA, Ghorbaninejad, Z, Piryaei, A, and Baharvand, H (2020). Organoid technology in female reproductive biomedicine. Reproductive Biology and Endocrinology 18, 64.
Organoid technology in female reproductive biomedicine.Crossref | GoogleScholarGoogle Scholar | 32552764PubMed |

Hemler, ME (2005). Tetraspanin functions and associated microdomains. Nature Reviews. Molecular Cell Biology 6, 801–811.
Tetraspanin functions and associated microdomains.Crossref | GoogleScholarGoogle Scholar | 16314869PubMed |

Higuchi, C, Nakatsuka, A, Eguchi, J, Teshigawara, S, Kanzaki, M, Katayama, A, et al. (2015). Identification of circulating miR-101, miR-375 and miR-802 as biomarkers for type 2 diabetes. Metabolism: Clinical and Experimental 64, 489–497.
Identification of circulating miR-101, miR-375 and miR-802 as biomarkers for type 2 diabetes.Crossref | GoogleScholarGoogle Scholar |

Hoelker, M, Rings, F, Lund, Q, Phatsara, C, Schellander, K, and Tesfaye, D (2010). Effect of embryo density on in vitro developmental characteristics of bovine preimplantative embryos with respect to micro and macroenvironments. Reproduction in Domestic Animals 45, e138–145.
Effect of embryo density on in vitro developmental characteristics of bovine preimplantative embryos with respect to micro and macroenvironments.Crossref | GoogleScholarGoogle Scholar | 19912573PubMed |

Horibe, S, Tanahashi, T, Kawauchi, S, Murakami, Y, and Rikitake, Y (2018). Mechanism of recipient cell-dependent differences in exosome uptake. BMC Cancer 18, 47.
Mechanism of recipient cell-dependent differences in exosome uptake.Crossref | GoogleScholarGoogle Scholar | 29306323PubMed |

Hoshino, A, Costa-Silva, B, Shen, T-L, Rodrigues, G, Hashimoto, A, Tesic Mark, M, et al. (2015). Tumour exosome integrins determine organotropic metastasis. Nature 527, 329–335.
Tumour exosome integrins determine organotropic metastasis.Crossref | GoogleScholarGoogle Scholar | 26524530PubMed |

Hou, C-H, Lin, F-L, Hou, S-M, and Liu, J-F (2014). Hyperthermia induces apoptosis through endoplasmic reticulum and reactive oxygen species in human osteosarcoma cells. International Journal of Molecular Sciences 15, 17380–17395.
Hyperthermia induces apoptosis through endoplasmic reticulum and reactive oxygen species in human osteosarcoma cells.Crossref | GoogleScholarGoogle Scholar | 25268613PubMed |

Hu, J, Tang, T, Zeng, Z, Wu, J, Tan, X, and Yan, J (2020). The expression of small RNAs in exosomes of follicular fluid altered in human polycystic ovarian syndrome. PeerJ 8, e8640.
The expression of small RNAs in exosomes of follicular fluid altered in human polycystic ovarian syndrome.Crossref | GoogleScholarGoogle Scholar | 32117643PubMed |

Hung, W-T, Hong, X, Christenson, LK, and McGinnis, LK (2015). Extracellular vesicles from bovine follicular fluid support cumulus expansion. Biology of Reproduction 93, 117.
Extracellular vesicles from bovine follicular fluid support cumulus expansion.Crossref | GoogleScholarGoogle Scholar | 26423123PubMed |

Hung, W-T, Navakanitworakul, R, Khan, T, Zhang, P, Davis, JS, McGinnis, LK, and Christenson, LK (2017). Stage-specific follicular extracellular vesicle uptake and regulation of bovine granulosa cell proliferation. Biology of Reproduction 97, 644–655.
Stage-specific follicular extracellular vesicle uptake and regulation of bovine granulosa cell proliferation.Crossref | GoogleScholarGoogle Scholar | 29025042PubMed |

Ibrahim, A, and Marbán, E (2016). Exosomes: fundamental biology and roles in cardiovascular physiology. Annual Review of Physiology 78, 67–83.
Exosomes: fundamental biology and roles in cardiovascular physiology.Crossref | GoogleScholarGoogle Scholar | 26667071PubMed |

Jacobs, LA, Bewicke-Copley, F, Poolman, MG, Pink, RC, Mulcahy, LA, Baker, I, et al. (2013). Meta-analysis using a novel database, miRStress, reveals miRNAs that are frequently associated with the radiation and hypoxia stress-responses. PLoS ONE 8, e80844.
Meta-analysis using a novel database, miRStress, reveals miRNAs that are frequently associated with the radiation and hypoxia stress-responses.Crossref | GoogleScholarGoogle Scholar | 24244721PubMed |

Jiang, X, Li, J, Zhang, B, Hu, J, Ma, J, Cui, L, and Chen, Z-J (2021). Differential expression profile of plasma exosomal microRNAs in women with polycystic ovary syndrome. Fertility and Sterility 115, 782–792.
Differential expression profile of plasma exosomal microRNAs in women with polycystic ovary syndrome.Crossref | GoogleScholarGoogle Scholar | 33041053PubMed |

Kalluri, R, and LeBleu, VS (2020). The biology, function, and biomedical applications of exosomes. Science (New York, NY) 367, eaau6977.
The biology, function, and biomedical applications of exosomes.Crossref | GoogleScholarGoogle Scholar |

Keller, S, Ridinger, J, Rupp, A-K, Janssen, JWG, and Altevogt, P (2011). Body fluid derived exosomes as a novel template for clinical diagnostics. Journal of Translational Medicine 9, 86.
Body fluid derived exosomes as a novel template for clinical diagnostics.Crossref | GoogleScholarGoogle Scholar | 21651777PubMed |

Kim, K, Mitsumizo, N, Fujita, K, and Utsumi, K (1996). The effects of follicular fluid on in vitro maturation, oocyte fertilization and the development of bovine embryos. Theriogenology 45, 787–799.
The effects of follicular fluid on in vitro maturation, oocyte fertilization and the development of bovine embryos.Crossref | GoogleScholarGoogle Scholar | 16727841PubMed |

Kirkman, MS, Briscoe, VJ, Clark, N, Florez, H, Haas, LB, Halter, JB, et al. (2012). Diabetes in older adults. Diabetes Care 35, 2650–2664.
Diabetes in older adults.Crossref | GoogleScholarGoogle Scholar | 23100048PubMed |

Kong, L, Zhu, J, Han, W, Jiang, X, Xu, M, Zhao, Y, et al. (2011). Significance of serum microRNAs in pre-diabetes and newly diagnosed type 2 diabetes: a clinical study. Acta Diabetologica 48, 61–69.
Significance of serum microRNAs in pre-diabetes and newly diagnosed type 2 diabetes: a clinical study.Crossref | GoogleScholarGoogle Scholar | 20857148PubMed |

Kosaka, N, Yoshioka, Y, Hagiwara, K, Tominaga, N, Katsuda, T, and Ochiya, T (2013). Trash or treasure: extracellular microRNAs and cell-to-cell communication. Frontiers in Genetics 4, 173.
Trash or treasure: extracellular microRNAs and cell-to-cell communication.Crossref | GoogleScholarGoogle Scholar | 24046777PubMed |

Laflamme, J, Akoum, A, and Leclerc, P (2005). Induction of human sperm capacitation and protein tyrosine phosphorylation by endometrial cells and interleukin-6. Molecular Human Reproduction 11, 141–150.
Induction of human sperm capacitation and protein tyrosine phosphorylation by endometrial cells and interleukin-6.Crossref | GoogleScholarGoogle Scholar | 15665187PubMed |

Lai, CP, Kim, EY, Badr, CE, Weissleder, R, Mempel, TR, Tannous, BA, and Breakefield, XO (2015). Visualization and tracking of tumour extracellular vesicle delivery and RNA translation using multiplexed reporters. Nature Communications 6, 7029.
Visualization and tracking of tumour extracellular vesicle delivery and RNA translation using multiplexed reporters.Crossref | GoogleScholarGoogle Scholar | 25967391PubMed |

Lancaster, GI, and Febbraio, MA (2005). Exosome-dependent trafficking of HSP70: a novel secretory pathway for cellular stress proteins. The Journal of Biological Chemistry 280, 23349–23355.
Exosome-dependent trafficking of HSP70: a novel secretory pathway for cellular stress proteins.Crossref | GoogleScholarGoogle Scholar | 15826944PubMed |

Lancaster, MA, and Knoblich, JA (2014). Organogenesis in a dish: modeling development and disease using organoid technologies. Science (New York, NY) 345, 1247125.
Organogenesis in a dish: modeling development and disease using organoid technologies.Crossref | GoogleScholarGoogle Scholar |

Leese, HJ, Hugentobler, SA, Gray, SM, Morris, DG, Sturmey, RG, Whitear, S-L, and Sreenan, JM (2008). Female reproductive tract fluids: composition, mechanism of formation and potential role in the developmental origins of health and disease. Reproduction, Fertility, and Development 20, 1–8.
Female reproductive tract fluids: composition, mechanism of formation and potential role in the developmental origins of health and disease.Crossref | GoogleScholarGoogle Scholar | 18154692PubMed |

Leroy, JLMR, Vanholder, T, Delanghe, JR, Opsomer, G, van Soom, A, Bols, PEJ, et al. (2004). Metabolic changes in follicular fluid of the dominant follicle in high-yielding dairy cows early post partum. Theriogenology 62, 1131–1143.
Metabolic changes in follicular fluid of the dominant follicle in high-yielding dairy cows early post partum.Crossref | GoogleScholarGoogle Scholar |

Li, J, Gao, H, Tian, Z, Wu, Y, Wang, Y, Fang, Y, et al. (2016). Effects of chronic heat stress on granulosa cell apoptosis and follicular atresia in mouse ovary. Journal of Animal Science and Biotechnology 7, 57.
Effects of chronic heat stress on granulosa cell apoptosis and follicular atresia in mouse ovary.Crossref | GoogleScholarGoogle Scholar | 27708774PubMed |

Liang, B, Peng, P, Chen, S, Li, L, Zhang, M, Cao, D, et al. (2013). Characterization and proteomic analysis of ovarian cancer-derived exosomes. Journal of Proteomics 80, 171–182.
Characterization and proteomic analysis of ovarian cancer-derived exosomes.Crossref | GoogleScholarGoogle Scholar | 23333927PubMed |

Lopera-Vásquez, R, Hamdi, M, Fernandez-Fuertes, B, Maillo, V, Beltrán-Breña, P, Calle, A, et al. (2016). Extracellular vesicles from BOEC in in vitro embryo development and quality. PLoS ONE 11, e0148083.
Extracellular vesicles from BOEC in in vitro embryo development and quality.Crossref | GoogleScholarGoogle Scholar | 26845570PubMed |

Lopera-Vasquez, R, Hamdi, Me, Maillo, V, Gutierrez-Adan, A, Bermejo-Alvarez, P, Ramírez, MÁ, et al. (2017). Effect of bovine oviductal extracellular vesicles on embryo development and quality in vitro. Reproduction (Cambridge, England) 153, 461–470.
Effect of bovine oviductal extracellular vesicles on embryo development and quality in vitro.Crossref | GoogleScholarGoogle Scholar |

López-Otín, C, Blasco, MA, Partridge, L, Serrano, M, and Kroemer, G (2013). The hallmarks of aging. Cell 153, 1194–1217.
The hallmarks of aging.Crossref | GoogleScholarGoogle Scholar | 23746838PubMed |

Lucy, MC (2001). Reproductive loss in high-producing dairy cattle: where will it end? In Journal of Dairy Science 84, 1277–1293.
Reproductive loss in high-producing dairy cattle: where will it end?Crossref | GoogleScholarGoogle Scholar |

Lv, A, Tu, Z, Huang, Y, Lu, W, and Xie, B (2021). Circulating exosomal miR-125a-5p as a novel biomarker for cervical cancer. Oncology Letters 21, 54.
Circulating exosomal miR-125a-5p as a novel biomarker for cervical cancer.Crossref | GoogleScholarGoogle Scholar | 33281965PubMed |

Machtinger, R, Laurent, LC, and Baccarelli, AA (2016). Extracellular vesicles: roles in gamete maturation, fertilization and embryo implantation. Human Reproduction Update 22, 182–193.
Extracellular vesicles: roles in gamete maturation, fertilization and embryo implantation.Crossref | GoogleScholarGoogle Scholar | 26663221PubMed |

María, Y-M, Pia, R-MS, Zoraida, A, Apolonija, BZ, Francesc, EB, Edit, IB, et al. (2015). Biological properties of extracellular vesicles and their physiological functions. Journal of Extracellular Vesicles 4, 27066.
Biological properties of extracellular vesicles and their physiological functions.Crossref | GoogleScholarGoogle Scholar |

Mathieu, M, Martin-Jaular, L, Lavieu, G, and Théry, C (2019). Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nature Cell Biology 21, 9–17.
Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication.Crossref | GoogleScholarGoogle Scholar | 30602770PubMed |

Meuwissen, TH, Hayes, BJ, and Goddard, ME (2001). Prediction of total genetic value using genome-wide dense marker maps. Genetics 157, 1819–1829.
| 11290733PubMed |

Montecalvo, A, Larregina, AT, Shufesky, WJ, Stolz, DB, Sullivan, MLG, Karlsson, JM, et al. (2012). Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes. Blood 119, 756–766.
Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes.Crossref | GoogleScholarGoogle Scholar | 22031862PubMed |

Morelli, AE, Larregina, AT, Shufesky, WJ, Sullivan, MLG, Stolz, DB, Papworth, GD, et al. (2004). Endocytosis, intracellular sorting, and processing of exosomes by dendritic cells. Blood 104, 3257–3266.
Endocytosis, intracellular sorting, and processing of exosomes by dendritic cells.Crossref | GoogleScholarGoogle Scholar | 15284116PubMed |

Mulcahy, LA, Pink, RC, and Carter, DRF (2014). Routes and mechanisms of extracellular vesicle uptake. Journal of Extracellular Vesicles 3, 24641.
Routes and mechanisms of extracellular vesicle uptake.Crossref | GoogleScholarGoogle Scholar |

Murdica, V, Giacomini, E, Makieva, S, Zarovni, N, Candiani, M, Salonia, A, et al. (2020). In vitro cultured human endometrial cells release extracellular vesicles that can be uptaken by spermatozoa. Scientific Reports 10, 8856.
In vitro cultured human endometrial cells release extracellular vesicles that can be uptaken by spermatozoa.Crossref | GoogleScholarGoogle Scholar | 32483153PubMed |

Nabenishi, H, Ohta, H, Nishimoto, T, Morita, T, Ashizawa, K, and Tsuzuki, Y (2012). The effects of cysteine addition during in vitro maturation on the developmental competence, ROS, GSH and apoptosis level of bovine oocytes exposed to heat stress. Zygote (Cambridge, England) 20, 249–259.
The effects of cysteine addition during in vitro maturation on the developmental competence, ROS, GSH and apoptosis level of bovine oocytes exposed to heat stress.Crossref | GoogleScholarGoogle Scholar |

Ng, YH, Rome, S, Jalabert, A, Forterre, A, Singh, H, Hincks, CL, and Salamonsen, LA (2013). Endometrial exosomes/microvesicles in the uterine microenvironment: a new paradigm for embryo-endometrial cross talk at implantation. PLoS ONE 8, e58502.
Endometrial exosomes/microvesicles in the uterine microenvironment: a new paradigm for embryo-endometrial cross talk at implantation.Crossref | GoogleScholarGoogle Scholar | 23516492PubMed |

O’Doherty, EM, Wade, MG, Hill, JL, and Boland, MP (1997). Effects of culturing bovine oocytes either singly or in groups on development to blastocysts. Theriogenology 48, 161–169.
Effects of culturing bovine oocytes either singly or in groups on development to blastocysts.Crossref | GoogleScholarGoogle Scholar | 16728116PubMed |

Ofir-Birin, Y, Abou Karam, P, Rudik, A, Giladi, T, Porat, Z, and Regev-Rudzki, N (2018). Monitoring extracellular vesicle cargo active uptake by imaging flow cytometry. Frontiers in Immunology 9, 1011.
Monitoring extracellular vesicle cargo active uptake by imaging flow cytometry.Crossref | GoogleScholarGoogle Scholar | 29881375PubMed |

O’Neill, C (2008). The potential roles for embryotrophic ligands in preimplantation embryo development. Human Reproduction Update 14, 275–288.
| 18281694PubMed |

O’Shea, LC, Mehta, J, Lonergan, P, Hensey, C, and Fair, T (2012). Developmental competence in oocytes and cumulus cells: candidate genes and networks. Systems Biology in Reproductive Medicine 58, 88–101.
Developmental competence in oocytes and cumulus cells: candidate genes and networks.Crossref | GoogleScholarGoogle Scholar | 22313243PubMed |

Ostrowski, M, Carmo, NB, Krumeich, S, Fanget, I, Raposo, G, Savina, A, et al. (2010). Rab27a and Rab27b control different steps of the exosome secretion pathway. Nature Cell Biology 12, 19–30.
Rab27a and Rab27b control different steps of the exosome secretion pathway.Crossref | GoogleScholarGoogle Scholar | 19966785PubMed |

Pavani, KC, Hendrix, A, van den Broeck, W, Couck, L, Szymanska, K, Lin, X, et al. (2019). Isolation and characterization of functionally active extracellular vesicles from culture medium conditioned by bovine embryos in vitro. International Journal of Molecular Sciences 20, .
Isolation and characterization of functionally active extracellular vesicles from culture medium conditioned by bovine embryos in vitro.Crossref | GoogleScholarGoogle Scholar |

Poirier, M, Tesfaye, D, Hailay, T, Salilew-Wondim, D, Gebremedhn, S, Rings, F, et al. (2020). Metabolism-associated genome-wide epigenetic changes in bovine oocytes during early lactation. Scientific Reports 10, 2345.
Metabolism-associated genome-wide epigenetic changes in bovine oocytes during early lactation.Crossref | GoogleScholarGoogle Scholar | 32047242PubMed |

Pontes, JHF, Nonato-Junior, I, Sanches, BV, Ereno-Junior, JC, Uvo, S, Barreiros, TRR, et al. (2009). Comparison of embryo yield and pregnancy rate between in vivo and in vitro methods in the same Nelore (Bos indicus) donor cows. Theriogenology 71, 690–697.
Comparison of embryo yield and pregnancy rate between in vivo and in vitro methods in the same Nelore (Bos indicus) donor cows.Crossref | GoogleScholarGoogle Scholar |

Qu, P, Luo, S, Du, Y, Zhang, Y, Song, X, Yuan, X, et al. (2020). Extracellular vesicles and melatonin benefit embryonic develop by regulating reactive oxygen species and 5-methylcytosine. Journal of Pineal Research 68, e12635.
Extracellular vesicles and melatonin benefit embryonic develop by regulating reactive oxygen species and 5-methylcytosine.Crossref | GoogleScholarGoogle Scholar | 32012354PubMed |

Racicot, K, Schmitt, A, and Ott, T (2012). The myxovirus-resistance protein, MX1, is a component of exosomes secreted by uterine epithelial cells. American Journal of Reproductive Immunology 67, 498–505.
The myxovirus-resistance protein, MX1, is a component of exosomes secreted by uterine epithelial cells.Crossref | GoogleScholarGoogle Scholar | 22574859PubMed |

Raposo, G, and Stoorvogel, W (2013). Extracellular vesicles: exosomes, microvesicles, and friends. The Journal of Cell Biology 200, 373–383.
Extracellular vesicles: exosomes, microvesicles, and friends.Crossref | GoogleScholarGoogle Scholar | 23420871PubMed |

Rivera, RM, and Hansen, PJ (2001). Development of cultured bovine embryos after exposure to high temperatures in the physiological range. Reproduction (Cambridge, England) 121, 107–115.

Rodrigues, TA, Tuna, KM, Alli, AA, Tribulo, P, Hansen, PJ, Koh, J, and Paula-Lopes, FF (2019). Follicular fluid exosomes act on the bovine oocyte to improve oocyte competence to support development and survival to heat shock. Reproduction, Fertility, and Development 31, 888–897.
Follicular fluid exosomes act on the bovine oocyte to improve oocyte competence to support development and survival to heat shock.Crossref | GoogleScholarGoogle Scholar | 30760387PubMed |

Romero-Arredondo, A, and Seidel, GE (1996). Effects of follicular fluid during in vitro maturation of bovine oocytes on in vitro fertilization and early embryonic development. Biology of Reproduction 55, 1012–1016.
Effects of follicular fluid during in vitro maturation of bovine oocytes on in vitro fertilization and early embryonic development.Crossref | GoogleScholarGoogle Scholar | 8902211PubMed |

Rosas-Hernandez, H, Chigurupati, S, Raymick, J, Robinson, B, Cuevas, E, Hanig, J, and Sarkar, S (2018). Identification of altered microRNAs in serum of a mouse model of Parkinson’s disease. Neuroscience Letters 687, 1–9.
Identification of altered microRNAs in serum of a mouse model of Parkinson’s disease.Crossref | GoogleScholarGoogle Scholar | 30025832PubMed |

Roth, Z (2017). Effect of heat stress on reproduction in dairy cows: insights into the cellular and molecular responses of the oocyte. Annual Review of Animal Biosciences 5, 151–170.
Effect of heat stress on reproduction in dairy cows: insights into the cellular and molecular responses of the oocyte.Crossref | GoogleScholarGoogle Scholar | 27732786PubMed |

Roth, Z, and Hansen, PJ (2005). Disruption of nuclear maturation and rearrangement of cytoskeletal elements in bovine oocytes exposed to heat shock during maturation. Reproduction (Cambridge, England) 129, 235–244.
Disruption of nuclear maturation and rearrangement of cytoskeletal elements in bovine oocytes exposed to heat shock during maturation.Crossref | GoogleScholarGoogle Scholar |

Roth, Z, and Wolfenson, D (2016). Comparing the effects of heat stress and mastitis on ovarian function in lactating cows: basic and applied aspects. Domestic Animal Endocrinology 56, S218–S227.
Comparing the effects of heat stress and mastitis on ovarian function in lactating cows: basic and applied aspects.Crossref | GoogleScholarGoogle Scholar | 27345320PubMed |

Roth, Z, Meidan, R, Shaham-Albalancy, A, Braw-Tal, R, and Wolfenson, D (2001). Delayed effect of heat stress on steroid production in medium-sized and preovulatory bovine follicles. Reproduction (Cambridge, England) 121, 745–751.

Ruiz-González, I, Xu, J, Wang, X, Burghardt, RC, Dunlap, KA, and Bazer, FW (2015). Exosomes, endogenous retroviruses and toll-like receptors: pregnancy recognition in ewes. Reproduction (Cambridge, England) 149, 281–291.
Exosomes, endogenous retroviruses and toll-like receptors: pregnancy recognition in ewes.Crossref | GoogleScholarGoogle Scholar |

Rukkwamsuk, T, Geelen, MJ, Kruip, TA, and Wensing, T (2000). Interrelation of fatty acid composition in adipose tissue, serum, and liver of dairy cows during the development of fatty liver postpartum. Journal of Dairy Science 83, 52–59.
Interrelation of fatty acid composition in adipose tissue, serum, and liver of dairy cows during the development of fatty liver postpartum.Crossref | GoogleScholarGoogle Scholar | 10659963PubMed |

Saadeldin, IM, Kim, SJ, Choi, YB, and Lee, BC (2014). Improvement of cloned embryos development by co-culturing with parthenotes: a possible role of exosomes/microvesicles for embryos paracrine communication. Cellular Reprogramming 16, 223–234.
Improvement of cloned embryos development by co-culturing with parthenotes: a possible role of exosomes/microvesicles for embryos paracrine communication.Crossref | GoogleScholarGoogle Scholar | 24773308PubMed |

Saeed-Zidane, M, Linden, L, Salilew-Wondim, D, Held, E, Neuhoff, C, Tholen, E, et al. (2017). Cellular and exosome mediated molecular defense mechanism in bovine granulosa cells exposed to oxidative stress. PLoS ONE 12, e0187569.
Cellular and exosome mediated molecular defense mechanism in bovine granulosa cells exposed to oxidative stress.Crossref | GoogleScholarGoogle Scholar | 29117219PubMed |

Santonocito, M, Vento, M, Guglielmino, MR, Battaglia, R, Wahlgren, J, Ragusa, M, et al. (2014). Molecular characterization of exosomes and their microRNA cargo in human follicular fluid: bioinformatic analysis reveals that exosomal microRNAs control pathways involved in follicular maturation. Fertility and Sterility 102, 1751–1761.e1.
Molecular characterization of exosomes and their microRNA cargo in human follicular fluid: bioinformatic analysis reveals that exosomal microRNAs control pathways involved in follicular maturation.Crossref | GoogleScholarGoogle Scholar | 25241362PubMed |

Senbon, S, Hirao, Y, and Miyano, T (2003). Interactions between the oocyte and surrounding somatic cells in follicular development: lessons from in vitro culture. The Journal of Reproduction and Development 49, 259–269.
Interactions between the oocyte and surrounding somatic cells in follicular development: lessons from in vitro culture.Crossref | GoogleScholarGoogle Scholar | 14967918PubMed |

Shkolnik, K, Tadmor, A, Ben-Dor, S, Nevo, N, Galiani, D, and Dekel, N (2011). Reactive oxygen species are indispensable in ovulation. Proceedings of the National Academy of Sciences of the United States of America 108, 1462–1467.
Reactive oxygen species are indispensable in ovulation.Crossref | GoogleScholarGoogle Scholar | 21220312PubMed |

Sidrat, T, Khan, AA, Joo, M-D, Wei, Y, Lee, K-L, Xu, L, and Kong, Il-K (2020). Bovine oviduct epithelial cell-derived culture media and exosomes improve mitochondrial health by restoring metabolic flux during pre-implantation development. International Journal of Molecular Sciences 21, 7589.
Bovine oviduct epithelial cell-derived culture media and exosomes improve mitochondrial health by restoring metabolic flux during pre-implantation development.Crossref | GoogleScholarGoogle Scholar |

Simon, C, Greening, DW, Bolumar, D, Balaguer, N, Salamonsen, LA, and Vilella, F (2018). Extracellular vesicles in human reproduction in health and disease. Endocrine Reviews 39, 292–332.
Extracellular vesicles in human reproduction in health and disease.Crossref | GoogleScholarGoogle Scholar | 29390102PubMed |

Sohel, MMH, Hoelker, M, Noferesti, SS, Salilew-Wondim, D, Tholen, E, Looft, C, et al. (2013). Exosomal and non-exosomal transport of extra-cellular micrornas in follicular fluid: implications for bovine oocyte developmental competence. PLoS ONE 8, e78505.
Exosomal and non-exosomal transport of extra-cellular micrornas in follicular fluid: implications for bovine oocyte developmental competence.Crossref | GoogleScholarGoogle Scholar |

Somfai, T, Inaba, Y, Watanabe, S, Geshi, M, and Nagai, T (2012). Follicular fluid supplementation during in vitro maturation promotes sperm penetration in bovine oocytes by enhancing cumulus expansion and increasing mitochondrial activity in oocytes. Reproduction, Fertility, and Development 24, 743–752.
Follicular fluid supplementation during in vitro maturation promotes sperm penetration in bovine oocytes by enhancing cumulus expansion and increasing mitochondrial activity in oocytes.Crossref | GoogleScholarGoogle Scholar | 22697124PubMed |

Stephen, JG, and Graça, R (2013). As we wait: coping with an imperfect nomenclature for extracellular vesicles. Journal of Extracellular Vesicles 2, 20389.
As we wait: coping with an imperfect nomenclature for extracellular vesicles.Crossref | GoogleScholarGoogle Scholar |

Stokes, PJ, Abeydeera, LR, and Leese, HJ (2005). Development of porcine embryos in vivo and in vitro; evidence for embryo ‘cross talk’ in vitro. Developmental Biology 284, 62–71.
Development of porcine embryos in vivo and in vitro; evidence for embryo ‘cross talk’ in vitro.Crossref | GoogleScholarGoogle Scholar | 15963973PubMed |

Sullivan, R, and Saez, F (2013). Epididymosomes, prostasomes, and liposomes: their roles in mammalian male reproductive physiology. Reproduction (Cambridge, England) 146, R21–R35.
Epididymosomes, prostasomes, and liposomes: their roles in mammalian male reproductive physiology.Crossref | GoogleScholarGoogle Scholar |

Sutton, ML, Gilchrist, RB, and Thompson, JG (2003). Effects of in-vivo and in-vitro environments on the metabolism of the cumulus-oocyte complex and its influence on oocyte developmental capacity. Human Reproduction Update 9, 35–48.
Effects of in-vivo and in-vitro environments on the metabolism of the cumulus-oocyte complex and its influence on oocyte developmental capacity.Crossref | GoogleScholarGoogle Scholar | 12638780PubMed |

Taka, M, Iwayama, H, and Fukui, Y (2005). Effect of the well of the well (WOW) system on in vitro culture for porcine embryos after intracytoplasmic sperm injection. The Journal of Reproduction and Development 51, 533–537.
Effect of the well of the well (WOW) system on in vitro culture for porcine embryos after intracytoplasmic sperm injection.Crossref | GoogleScholarGoogle Scholar | 16141648PubMed |

Tang, K, Zhang, Y, Zhang, H, Xu, P, Liu, J, Ma, J, et al. (2012). Delivery of chemotherapeutic drugs in tumour cell-derived microparticles. Nature Communications 3, 1282.
Delivery of chemotherapeutic drugs in tumour cell-derived microparticles.Crossref | GoogleScholarGoogle Scholar | 23250412PubMed |

Théry, C (2015). Cancer: diagnosis by extracellular vesicles. Nature 523, 161–162.
Cancer: diagnosis by extracellular vesicles.Crossref | GoogleScholarGoogle Scholar | 26106856PubMed |

Tkach, M, and Théry, C (2016). Communication by extracellular vesicles: where we are and where we need to go. Cell 164, 1226–1232.
Communication by extracellular vesicles: where we are and where we need to go.Crossref | GoogleScholarGoogle Scholar | 26967288PubMed |

Vajta, G, Korösi, T, Du, Y, Nakata, K, Ieda, S, Kuwayama, M, and Nagy, ZP (2008). The well-of-the-well system: an efficient approach to improve embryo development. Reproductive Biomedicine Online 17, 73–81.
The well-of-the-well system: an efficient approach to improve embryo development.Crossref | GoogleScholarGoogle Scholar | 18616894PubMed |

Valadi, H, Ekström, K, Bossios, A, Sjöstrand, M, Lee, JJ, and Lötvall, JO (2007). Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature Cell Biology 9, 654–659.
Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells.Crossref | GoogleScholarGoogle Scholar | 17486113PubMed |

Vanholder, T, Leroy, JLMR, van Soom, A, Opsomer, G, Maes, D, Coryn, M, and de Kruif, A (2005). Effect of non-esterified fatty acids on bovine granulosa cell steroidogenesis and proliferation in vitro. Animal Reproduction Science 87, 33–44.
Effect of non-esterified fatty acids on bovine granulosa cell steroidogenesis and proliferation in vitro.Crossref | GoogleScholarGoogle Scholar | 15885439PubMed |

Vilella, F, Moreno-Moya, JM, Balaguer, N, Grasso, A, Herrero, M, Martínez, S, et al. (2015). Hsa-miR-30d, secreted by the human endometrium, is taken up by the pre-implantation embryo and might modify its transcriptome. Development 142, 3210–3221.
Hsa-miR-30d, secreted by the human endometrium, is taken up by the pre-implantation embryo and might modify its transcriptome.Crossref | GoogleScholarGoogle Scholar | 26395145PubMed |

Villarroya-Beltri, C, Baixauli, F, Gutiérrez-Vázquez, C, Sánchez-Madrid, F, and Mittelbrunn, M (2014). Sorting it out: regulation of exosome loading. Seminars in Cancer Biology 28, 3–13.
Sorting it out: regulation of exosome loading.Crossref | GoogleScholarGoogle Scholar | 24769058PubMed |

Visconti, PE, and Kopf, GS (1998). Regulation of protein phosphorylation during sperm capacitation. Biology of Reproduction 59, 1–6.
Regulation of protein phosphorylation during sperm capacitation.Crossref | GoogleScholarGoogle Scholar | 9674985PubMed |

Walters, AH, Pryor, AW, Bailey, TL, Pearson, RE, and Gwazdauskas, FC (2002). Milk yield, energy balance, hormone, follicular and oocyte measures in early and mid-lactation Holstein cows. Theriogenology 57, 949–961.
Milk yield, energy balance, hormone, follicular and oocyte measures in early and mid-lactation Holstein cows.Crossref | GoogleScholarGoogle Scholar | 11991397PubMed |

Wolfenson, D, Lew, BJ, Thatcher, WW, Graber, Y, and Meidan, R (1997). Seasonal and acute heat stress effects on steroid production by dominant follicles in cows. Animal Reproduction Science 47, 9–19.
Seasonal and acute heat stress effects on steroid production by dominant follicles in cows.Crossref | GoogleScholarGoogle Scholar | 9233502PubMed |

Wydooghe, E, Vandaele, L, Heras, S, de Sutter, P, Deforce, D, Peelman, L, et al. (2017). Autocrine embryotropins revisited: how do embryos communicate with each other in vitro when cultured in groups? Biological Reviews 92, 505–520.
Autocrine embryotropins revisited: how do embryos communicate with each other in vitro when cultured in groups?Crossref | GoogleScholarGoogle Scholar | 26608222PubMed |

Zhan, R, Leng, X, Liu, X, Wang, X, Gong, J, Yan, L, et al. (2009). Heat shock protein 70 is secreted from endothelial cells by a non-classical pathway involving exosomes. Biochemical and Biophysical Research Communications 387, 229–233.
Heat shock protein 70 is secreted from endothelial cells by a non-classical pathway involving exosomes.Crossref | GoogleScholarGoogle Scholar | 19555663PubMed |

Zheng, Y, Chen, K, Zheng, X, Li, H, and Wang, G (2014). Identification and bioinformatics analysis of microRNAs associated with stress and immune response in serum of heat-stressed and normal Holstein cows. Cell Stress & Chaperones 19, 973–981.
Identification and bioinformatics analysis of microRNAs associated with stress and immune response in serum of heat-stressed and normal Holstein cows.Crossref | GoogleScholarGoogle Scholar |